Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 51
Filtrar
1.
J Thromb Haemost ; 16(10): 2024-2034, 2018 10.
Artículo en Inglés | MEDLINE | ID: mdl-30070759

RESUMEN

Essentials Knowledge of genetic regulators of plasma factor VII activating protease (FSAP) levels is limited. We performed a genome-wide analysis of variants influencing FSAP activity in Scandinavian cohorts. We replicated an association for Marburg-1 and identified an association for a HABP2 stop variant. We identified a novel locus near ADCY2 as a potential additional regulator of FSAP activity. SUMMARY: Background Factor VII-activating protease (FSAP) has roles in both coagulation and fibrinolysis. Recent data indicate its involvement in several other processes, such as vascular remodeling and inflammation. Plasma FSAP activity is highly variable among healthy individuals and, apart from the low-frequency missense variant Marburg-I (rs7080536) in the FSAP-encoding gene HABP2, determinants of this variation are unclear. Objectives To identify novel genetic variants within and outside of the HABP2 locus that influence circulating FSAP activity. Patients/Methods We performed an exploratory genome-wide association study (GWAS) on plasma FSAP activity amongst 3230 Swedish subjects. Directly genotyped rare variants were also analyzed with gene-based tests. Using GWAS, we confirmed the strong association between the Marburg-I variant and FSAP activity. HABP2 was also significant in the gene-based analysis, and remained significant after exclusion of Marburg-I carriers. This was attributable to a rare HABP2 stop variant (rs41292628). Carriers of this stop variant showed a similar reduction in FSAP activity as Marburg-I carriers, and this finding was replicated. A secondary genome-wide significant locus was identified at a 5p15 locus (rs35510613), and this finding requires future replication. This common variant is located upstream of ADCY2, which encodes a protein catalyzing the formation of cAMP. Results and Conclusions This study verified the Marburg-I variant to be a strong regulator of FSAP activity, and identified an HABP2 stop variant with a similar impact on FSAP activity. A novel locus near ADCY2 was identified as a potential additional regulator of FSAP activity.


Asunto(s)
Adenilil Ciclasas/genética , Sitios Genéticos , Variación Genética , Hemostasis/genética , Serina Endopeptidasas/sangre , Serina Endopeptidasas/genética , Adolescente , Adulto , Anciano , Animales , Células Cultivadas , Femenino , Estudio de Asociación del Genoma Completo , Genotipo , Hepatocitos/enzimología , Humanos , Masculino , Ratones Endogámicos BALB C , Persona de Mediana Edad , Suecia , Adulto Joven
2.
J Thromb Haemost ; 14(10): 2058-2067, 2016 10.
Artículo en Inglés | MEDLINE | ID: mdl-27431088

RESUMEN

Essentials Factor VII-activating protease (FSAP) is a plasma protease involved in vascular processes. Neointima formation was investigated after vascular injury in FSAP-/- mice. The neointimal lesion size and the accumulation of macrophages were increased in FSAP-/- mice. This was due to an increased activity of the chemokine (C-C motif) ligand 2 (CCL2). SUMMARY: Background Factor VII-activating protease (FSAP) is a multifunctional circulating plasma serine protease involved in thrombosis and vascular remodeling processes. The Marburg I single-nucleotide polymorphism (MI-SNP) in the FSAP-coding gene is characterized by low proteolytic activity, and is associated with increased rates of stroke and carotid stenosis in humans. Objectives To determine whether neointima formation after vascular injury is increased in FSAP-/- mice. Methods and Results The neointimal lesion size and the proliferation of vascular smooth muscle cells (VSMCs) were significantly enhanced in FSAP-/- mice as compared with C57BL/6 control mice after wire-induced injury of the femoral artery. Accumulation of leukocytes and macrophages was increased within the lesions of FSAP-/- mice at day 3 and day 14. Quantitative zymography demonstrated enhanced activity of gelatinases/matrix metalloproteinase (MMP)-2 and MMP-9 within the neointimal lesions of FSAP-/- mice, and immunohistochemistry showed particular costaining of MMP-9 with accumulating leukocytes. Using intravital microscopy, we observed that FSAP deficiency promoted the intravascular adherence and the subsequent transmigration of leukocytes in vivo in response to chemokine ligand 2 (CCL2). CCL2 expression was increased in FSAP-/- monocytes but not in the vessel wall. There was no difference in the expression of platelet-derived growth factor (PDGF-BB). Conclusions FSAP deficiency causes an increase in CCL2 expression and CCL2-mediated infiltration of leukocytes into the injured vessel, thereby promoting SMC proliferation and migration by the activation of leukocyte-derived gelatinases. These results provide a possible explanation for the observed association of the loss-of-function MI-SNP with vascular proliferative diseases.


Asunto(s)
Leucocitos/citología , Neointima/sangre , Serina Endopeptidasas/deficiencia , Serina Endopeptidasas/genética , Animales , Becaplermina , Peso Corporal , Estenosis Carotídea , Movimiento Celular , Proliferación Celular , Quimiocina CCL2/genética , Quimiotaxis , Arteria Femoral/patología , Macrófagos/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Músculo Esquelético/patología , Miocitos del Músculo Liso/citología , Polimorfismo de Nucleótido Simple , Proteínas Proto-Oncogénicas c-sis/genética , Serina Endopeptidasas/sangre
3.
Eur J Neurosci ; 41(7): 965-75, 2015 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-25615590

RESUMEN

Factor VII activating protease (FSAP) is a circulating protease with a putative role in hemostasis, remodeling and inflammation. A polymorphism giving rise to low proteolytic activity has been associated with an increased risk of stroke and carotid stenosis. To date, no in vivo studies or mechanistic information is available to explain these results. Based on the polymorphism data we hypothesize that a lack of endogenous FSAP will increase the severity of stroke. Stroke was induced by applying thrombin in the middle cerebral artery in wild-type (WT) and FSAP(-/-) mice. Increased stroke volume and worsened neurological deficit were observed in FSAP(-/-) mice. Raised levels of FSAP protein were detected in the infarcted area of WT mice together with enhanced leukocyte infiltration and apoptosis in FSAP(-/-) mice. There was a concomitant increase in the activation of the NFκB pathway and decrease in expression of the PI3K/AKT pathway proteins. At a cellular level, FSAP increased cell survival and decreased apoptosis in primary cortical neurons and astrocytes exposed to tPA/NMDA excitotoxicity or oxygen glucose deprivation (OGD)/reoxygenation, respectively. This was mediated via the PI3K/AKT pathway with involvement of the protease activated receptor-1. To corroborate the human epidemiological data, which link FSAP with stroke, we now show that the lack of FSAP in mice worsens the outcome of stroke. In the absence of FSAP there was a stronger inflammatory response and lower cell survival due to insufficient activation of the PI3K/AKT pathway.


Asunto(s)
Isquemia Encefálica/enzimología , Serina Endopeptidasas/deficiencia , Accidente Cerebrovascular/enzimología , Animales , Apoptosis/fisiología , Astrocitos/enzimología , Astrocitos/patología , Encéfalo/enzimología , Encéfalo/patología , Isquemia Encefálica/patología , Movimiento Celular/fisiología , Supervivencia Celular/fisiología , Células Cultivadas , Modelos Animales de Enfermedad , Infarto de la Arteria Cerebral Media , Leucocitos/patología , Leucocitos/fisiología , Masculino , Ratones Endogámicos C57BL , Ratones Noqueados , Neuronas/enzimología , Neuronas/patología , Receptor PAR-1/metabolismo , Serina Endopeptidasas/genética , Liberación Accidental en Seveso , Accidente Cerebrovascular/patología , Trombina
4.
Thromb Res ; 130(6): 935-41, 2012 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-22906531

RESUMEN

INTRODUCTION: Factor VII activating protease (FSAP) is a plasma protease with FVII and pro-urokinase (pro-uPA) activating properties. A single nucleotide polymorphism (SNP) (Marburg I, MI) in the FSAP gene (HABP-2) leads to a low activity of the MI-FSAP towards pro-uPA, but supposedly not towards FVII and is described as a risk factor for athero-thrombosis and liver fibrosis. Recently we found, however, that FVII is an extremely poor substrate of FSAP and identified tissue factor pathway inhibitor (TFPI) as a novel substrate for FSAP. This prompted us to re-investigate the proteolytic activity profile of FSAP and to re-define its role in haemostasis. MATERIAL AND METHODS: Using purified protein and genotyped plasma samples, we systematically compared the activities of wild type (WT) and MI-FSAP towards natural plasma substrates. The influence of FSAP on coagulation was studied in prothrombin time assays. RESULTS: FSAP from homozygous MI-carriers has a general low proteolytic activity making this variant a natural "knock-down". In human plasma WT-FSAP, but not MI-FSAP, accelerated the extrinsic coagulation by inactivation of TFPI. The diminished ability of MI-FSAP to cleave TFPI is reflected by a positive correlation between the FSAP enzymatic activity and cleaved TFPI in the circulation. CONCLUSION: Most likely TFPI cleavage by WT-FSAP occurs in vivo and contributes to an elevated level of endogenous FVIIa. This may explain why MI-FSAP is not a clear indicator for deep vein thrombosis in population studies. The loss of the pro-fibrinolytic protective function of FSAP in carriers of the MI-SNP may account for the association of the MI-SNP with atherosclerosis and thromboembolic complications.


Asunto(s)
Coagulación Sanguínea/fisiología , Serina Endopeptidasas/sangre , Serina Endopeptidasas/genética , Coagulación Sanguínea/genética , Estudios de Cohortes , Factor VIIa/metabolismo , Genotipo , Humanos , Polimorfismo de Nucleótido Simple , Proteolisis , Factores de Riesgo
5.
J Thromb Haemost ; 10(5): 848-56, 2012 May.
Artículo en Inglés | MEDLINE | ID: mdl-22409238

RESUMEN

BACKGROUND: Factor VII-activating protease (FSAP) is a recently discovered plasma protease with a role in the regulation of hemostasis and vascular remodeling processes. Higher levels and activity of FSAP have been reported in patients with deep vein thrombosis, but there are no data on plasma FSAP in ischemic stroke (IS). OBJECTIVE: To investigate whether FSAP antigen levels and activity are associated with IS and/or etiologic subtypes of IS. PATIENTS AND METHODS: To assess the potential association between FSAP and IS, plasma FSAP antigen levels and activity were measured in 600 consecutive IS patients and 600 population-based controls from the case-control study the Sahlgrenska Academy Study on Ischemic Stroke (SAHLSIS). Blood sampling was performed in the acute phase and 3 months after the index stroke. FSAP was also investigated at the genetic level by genotyping of 33 single-nucleotide polymorphisms. RESULTS: Increased FSAP antigen level and activity, at both time-points, were independently associated with IS. Subtype analysis revealed similar associations for both FSAP measures, at both time-points, in all main IS subtypes. FSAP genotypes showed association with both FSAP plasma measurements, but not with IS. CONCLUSIONS: Increased plasma FSAP antigen levels and activity were associated with IS and all main etiologic subtypes, suggesting a possible role for FSAP in the pathophysiology of IS, irrespective of the underlying etiology.


Asunto(s)
Isquemia Encefálica/enzimología , Serina Endopeptidasas/sangre , Accidente Cerebrovascular/enzimología , Adulto , Anciano , Biomarcadores/sangre , Isquemia Encefálica/sangre , Isquemia Encefálica/epidemiología , Isquemia Encefálica/genética , Estudios de Casos y Controles , Femenino , Genotipo , Humanos , Modelos Logísticos , Masculino , Persona de Mediana Edad , Análisis Multivariante , Oportunidad Relativa , Fenotipo , Polimorfismo de Nucleótido Simple , Medición de Riesgo , Factores de Riesgo , Serina Endopeptidasas/genética , Accidente Cerebrovascular/sangre , Accidente Cerebrovascular/epidemiología , Accidente Cerebrovascular/genética , Suecia/epidemiología , Regulación hacia Arriba
6.
Hamostaseologie ; 31(3): 174-6, 177-8, 2011 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-21655671

RESUMEN

Factor VII activating protease (FSAP) is a circulating serine protease with high homology to fibrinolytic enzymes. A role in the regulation of coagulation and fibrinolysis is suspected based on in vitro studies demonstrating activation of FVII or pro-urokinase plasminogen activator (uPA). However, considering the paucity of any studies in animal models or any correlative studies in humans the role of FSAP in haemostasis remains unclear. In relation to vascular remodeling processes or inflammation it has been convincingly shown that FSAP interacts with growth factors as well as protease activated receptors (PAR). Against this sparse background there are a plethora of studies which have investigated the linkage of single nucleotide polymorphisms (SNP) in the FSAP gene (HABP2) to various diseases. The G534E SNP of FSAP is associated with a low proteolytic activity due to an amino acid exchange in the protease domain. This and other SNPs have been linked to carotid stenosis, stroke as well as thrombosis in the elderly and plaque calcification. These SNP analyses indicate an important role for FSAP in the regulation of the haemostasis system as well as fibroproliferative inflammatory processes.


Asunto(s)
Coagulación Sanguínea/genética , Fibrinólisis/genética , Predisposición Genética a la Enfermedad/genética , Hemostasis/genética , Polimorfismo de Nucleótido Simple/genética , Serina Endopeptidasas/genética , Trombosis/genética , Coagulación Sanguínea/inmunología , Fibrinólisis/inmunología , Hemostasis/inmunología , Humanos , Serina Endopeptidasas/inmunología , Trombosis/inmunología
7.
Hamostaseologie ; 31(3): 174-178, 2011.
Artículo en Inglés | MEDLINE | ID: mdl-29589358

RESUMEN

Factor VII activating protease (FSAP) is a circulating serine protease with high homology to fibrinolytic enzymes. A role in the regulation of coagulation and fibrinolysis is suspected based on in vitro studies demonstrating activation of FVII or pro-urokinase plasminogen activator (uPA). However, considering the paucity of any studies in animal models or any correlative studies in humans the role of FSAP in haemostasis remains unclear. In relation to vascular remodeling processes or inflammation it has been convincingly shown that FSAP interacts with growth factors as well as protease activated receptors (PAR). Against this sparse background there are a plethora of studies which have investigated the linkage of single nucleotide polymorphisms (SNP) in the FSAP gene (HABP2) to various diseases. The G534E SNP of FSAP is associated with a low proteolytic activity due to an amino acid exchange in the protease domain. This and other SNPs have been linked to carotid stenosis, stroke as well as thrombosis in the elderly and plaque calcification. These SNP analyses indicate an important role for FSAP in the regulation of the haemostasis system as well as fibroproliferative inflammatory processes.

8.
J Thromb Haemost ; 8(6): 1313-22, 2010 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-20180903

RESUMEN

BACKGROUND AND OBJECTIVE: Platelets are essential for hemostasis, and they cause resistance to fibrinolysis by tissue-type plasminogen activator. In contrast, platelets enhance fibrinolysis mediated by single-chain urokinase-type plasminogen activator (scu-PA). This study investigated the mechanism behind this profibrinolytic role of platelets. METHODS AND RESULTS: Platelets enhanced scu-PA activity, but not urokinase-type plasminogen activator (u-PA) activity, in plasma clot lysis and chromogenic assays. We established, using the non-cleavable scu-PA mutant (Lys158-->Glu) and protease inhibitors, that platelets increased activation to u-PA by a serine protease. Activation of scu-PA was platelet-dependent, even in plasma. It occurred in platelet-rich but not in platelet-poor plasma, as assessed by sodium dodecylsulfate polyacrylamide gel electrophoresis and zymography after addition of plasminogen activator inhibitor-1. Candidate proteases that are known to activate scu-PA and are present in platelet preparations were investigated. Factor VII activating protease was detected in platelet preparations by western blotting, but its inhibition by antibodies did not inhibit activation of scu-PA by platelets. Plasmin and plasma kallikrein both mimicked the platelet effect, but were distinguished by their responses to a range of inhibitors. Analysis of platelet-associated protease activity and the time course of scu-PA activation pointed towards plasminogen, and the data were consistent with a mechanism of reciprocal activation. The essential role of plasminogen was revealed using platelets from plasminogen-deficient mice, which could not activate scu-PA. Local plasminogen on platelet membranes was markedly more effective than solution-phase plasminogen in activation of scu-PA. CONCLUSIONS: Platelets enhance fibrinolysis by scu-PA through reciprocal activation of scu-PA and platelet-associated plasminogen, a system that is potentially important in the lysis of platelet-rich thrombi.


Asunto(s)
Plaquetas/fisiología , Fibrinólisis , Plasminógeno/fisiología , Western Blotting , Activación Enzimática , Ensayo de Inmunoadsorción Enzimática , Humanos , Activador de Plasminógeno de Tipo Uroquinasa
10.
FASEB J ; 17(9): 1174-6, 2003 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-12692088

RESUMEN

To define the role of the plasminogen activators (PAs) urokinase PA (uPA) and tissue PA (tPA) as well as the uPA receptor (uPAR) in arteriogenesis, we investigated their impact in a rabbit and mouse model of adaptive collateral artery growth. Collateral artery growth was induced by occlusion of the femoral artery in rabbit and wild-type (WT) mice and in mice with targeted inactivation of uPA (uPA-/-), tPA (tPA-/-), or uPAR (uPAR-/-). Northern blot results revealed a significant up-regulation of uPA but not uPAR or tPA in the early phase of arteriogenesis in rabbit and WT mice. This up-regulation on RNA level was followed by an increased protein level and enzymatic activity. Impaired perfusion recovery upon femoral artery ligation was observed by laser Doppler analysis in vivo in uPA-deficient mice but not in uPAR or tPA deficiency compared with WT mice. Immunohistochemical studies revealed an association of leukocyte infiltration with arteriogenesis in WT mice that was strongly reduced in uPA-/- but not in uPAR- or tPA-deficient mice. We conclude that arteriogenesis is promoted by an uPA-mediated infiltration of leukocytes that is not dependent on uPAR.


Asunto(s)
Arterias/crecimiento & desarrollo , Activador de Plasminógeno de Tipo Uroquinasa/fisiología , Animales , Arterias/citología , Movimiento Celular , Arteria Femoral/cirugía , Miembro Posterior/irrigación sanguínea , Leucocitos/fisiología , Ligadura , Ratones , Ratones Noqueados , Modelos Cardiovasculares , ARN Mensajero/biosíntesis , Conejos , Receptores de Superficie Celular/genética , Receptores de Superficie Celular/fisiología , Receptores del Activador de Plasminógeno Tipo Uroquinasa , Flujo Sanguíneo Regional , Activador de Tejido Plasminógeno/genética , Activador de Tejido Plasminógeno/fisiología , Activador de Plasminógeno de Tipo Uroquinasa/genética
11.
Biochem Soc Trans ; 30(2): 168-73, 2002 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-12023845

RESUMEN

Vascular cell adhesion and migration, proliferation or differentiation are cellular responses that are induced by haemostatic factors of the urokinase/plasminogen activation complex, but the respective underlying mechanisms are largely undefined. The direct and indirect contributions of the urokinase-type plasminogen activator receptor (uPAR) system in inflammatory processes, as they relate to recruitment of leukocytes, define novel functions and could serve as therapeutic targets for related vasculopathies. The presence of uPAR plays a crucial role in beta2-integrin-mediated adhesion of leukocytes; uPAR also directly mediates leukocyte adhesion to vitronectin, a multifunctional adhesion protein that is associated with the extracellular matrix. The latter process is inhibited by plasminogen activator inhibitor-1. Both beta2-integrin- and uPAR-dependent processes are activated by Zn2+ and are blocked by high-molecular-mass kininogen. Domain 5 of kininogen was identified, in particular, as an anti-adhesive component with a potent anti-inflammatory action in a peritonitis mouse model. In patients with acute myocardial infarction, elevated expression of uPAR on monocytes resulted in their increased adherence to the endothelium, which indicates a possible role of the uPAR system in monocyte recruitment to the infarcted area. Urokinase-type plasminogen activator was identified as a potent mitogen for vascular smooth muscle cells, an observation that was independent of the presence of uPAR and its proteolytic activity. Taken together, these results strongly suggest an essential role for the uPAR system in acute inflammation as well as in chronic degenerative vascular processes such as atherosclerosis. Targeting the uPAR system may allow specific therapeutic intervention in vascular pathologies.


Asunto(s)
Hemostasis/fisiología , Inflamación/sangre , Inflamación/etiología , Quininógenos/sangre , Receptores de Superficie Celular/sangre , Animales , Adhesión Celular/fisiología , Humanos , Inflamación/patología , Integrinas/fisiología , Quininógenos/química , Quininógenos/fisiología , Ratones , Modelos Biológicos , Monocitos/patología , Monocitos/fisiología , Músculo Liso Vascular/patología , Músculo Liso Vascular/fisiopatología , Infarto del Miocardio/sangre , Infarto del Miocardio/etiología , Infarto del Miocardio/patología , Inhibidor 1 de Activador Plasminogénico/fisiología , Receptores de Superficie Celular/fisiología , Receptores del Activador de Plasminógeno Tipo Uroquinasa , Activador de Plasminógeno de Tipo Uroquinasa/fisiología
12.
FASEB J ; 15(13): 2365-76, 2001 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-11689462

RESUMEN

Proteolytic cleavage of single-chain, high molecular weight kininogen (HK) by kallikrein releases the short-lived vasodilator bradykinin and leaves behind a two-chain, high molecular weight kininogen (HKa) reported to bind to the beta2-integrin Mac-1 (CR3, CD11b/CD18, alphaMbeta2) on neutrophils and exert antiadhesive properties by binding to the urokinase receptor (uPAR) and vitronectin. We define the molecular mechanisms for the antiadhesive effects of HK related to disruption of beta2-integrin-mediated cellular interactions in vitro and in vivo. In a purified system, HK and HKa inhibited the binding of soluble fibrinogen and ICAM-1 to immobilized Mac-1, but not the binding of ICAM-1 to immobilized LFA-1 (CD11a/CD18, alphaLbeta2). This inhibitory effect could be attributed to HK domain 5 and to a lesser degree to HK domain 3, consistent with the requirement of both domains for binding to Mac-1. Accordingly, HK, HKa, and domain 5 inhibited the adhesion of Mac-1 but not LFA-1-transfected K562 human erythroleukemic cells to ICAM-1. Moreover, adhesion of human monocytic cells to fibrinogen and to human endothelial cells was blocked by HK, HKa, and domain 5. By using peptides derived from HK domain 5, the sequences including amino acids H475-G497 (and to a lesser extent, G440-H455) were identified as responsible for the antiadhesive effect, which was independent of uPAR. Finally, administration of domain 5 into mice, followed by induction of thioglycollate-provoked peritonitis, decreased the recruitment of neutrophils by approximately 70% in this model of acute inflammation. Taken together, HKa (and particularly domain 5) specifically interacts with Mac-1 but not with LFA-1, thereby blocking Mac-1-dependent leukocyte adhesion to fibrinogen and endothelial cells in vitro and in vivo and serving as a novel endogenous regulator of leukocyte recruitment into the inflamed tissue.


Asunto(s)
Quininógeno de Alto Peso Molecular/farmacología , Leucocitos/efectos de los fármacos , Péptidos/farmacología , Secuencia de Aminoácidos , Animales , Anticuerpos Monoclonales/farmacología , Unión Competitiva/efectos de los fármacos , Adhesión Celular/efectos de los fármacos , Línea Celular , Modelos Animales de Enfermedad , Relación Dosis-Respuesta a Droga , Femenino , Fibrinógeno/metabolismo , Humanos , Molécula 1 de Adhesión Intercelular/metabolismo , Células K562 , Quininógeno de Alto Peso Molecular/química , Leucocitos/citología , Leucocitos/metabolismo , Antígeno-1 Asociado a Función de Linfocito/genética , Antígeno-1 Asociado a Función de Linfocito/inmunología , Antígeno-1 Asociado a Función de Linfocito/metabolismo , Antígeno de Macrófago-1/genética , Antígeno de Macrófago-1/inmunología , Antígeno de Macrófago-1/metabolismo , Ratones , Ratones Endogámicos , Datos de Secuencia Molecular , Infiltración Neutrófila/efectos de los fármacos , Peritonitis/inducido químicamente , Peritonitis/patología , Peritonitis/prevención & control , Plásmidos/genética , Tioglicolatos/administración & dosificación , Células U937
13.
J Biol Chem ; 276(31): 28946-53, 2001 Aug 03.
Artículo en Inglés | MEDLINE | ID: mdl-11501527

RESUMEN

The interaction between urokinase plasminogen activator (uPA) and its cellular receptor (uPAR) is a key event in cell surface-associated plasminogen activation, relevant for cell migration and invasion. In order to define receptor recognition sites for uPA, we have expressed uPAR fragments as fusion products with the minor coat protein on the surface of M13 bacteriophages. Sequence analysis of cDNA fragments encoding uPA-binding peptides indicated the existence of a composite uPA-binding structure including all three uPAR domains. This finding was confirmed by experiments using an overlapping 15-mer peptide array covering the entire uPAR molecule. Four regions within the uPAR sequence were found to directly bind to uPA: two distinct regions containing amino acids 13--20 and amino acids 74--84 of the uPAR domain I, and regions in the putative loop 3 of the domains II and III. All the uPA-binding fragments from the three domains were shown to have an agonistic effect on uPA binding to immobilized uPAR. Furthermore, uPAR-(154--176) increased uPAR-transfected BAF3-cell adhesion on vitronectin in the presence of uPA, whereas uPAR-(247--276) stimulated the cell adhesion both in the absence or presence of uPA. The latter fragment was also able to augment the binding of vitronectin to uPAR in a purified system, thereby mimicking the effect of uPA on this interaction. These results indicate that uPA binding can take place to particular part(s) on several uPAR molecules and that direct uPAR-uPAR contacts may contribute to receptor activation and ligand binding.


Asunto(s)
Receptores de Superficie Celular/química , Receptores de Superficie Celular/metabolismo , Activador de Plasminógeno de Tipo Uroquinasa/química , Activador de Plasminógeno de Tipo Uroquinasa/metabolismo , Secuencia de Aminoácidos , Sitios de Unión , Unión Competitiva , Cromatografía de Afinidad , ADN Complementario , Humanos , Cinética , Ligandos , Modelos Moleculares , Datos de Secuencia Molecular , Fragmentos de Péptidos/química , Fragmentos de Péptidos/aislamiento & purificación , Fragmentos de Péptidos/metabolismo , Conformación Proteica , Receptores de Superficie Celular/genética , Receptores del Activador de Plasminógeno Tipo Uroquinasa , Proteínas Recombinantes de Fusión/química , Proteínas Recombinantes de Fusión/metabolismo , Proteínas Recombinantes/química , Proteínas Recombinantes/metabolismo , Alineación de Secuencia , Vitronectina/sangre , Vitronectina/metabolismo
14.
Thromb Haemost ; 86(2): 686-93, 2001 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-11522023

RESUMEN

Urokinase-type plasminogen activator (uPA) and its cell surface-receptor (uPAR) regulate cellular functions linked to adhesion and migration and contribute to pericellular proteolysis in tissue remodelling processes. Soluble uPAR (suPAR) is present in the circulation, peritoneal and ascitic fluid and in the cystic fluid from ovarian cancer. We have investigated the origin and the vascular distribution of the soluble receptor, which accounts for 10-20% of the total receptor in vascular endothelial and smooth muscle cells. Phase separation analysis of the cell conditioned media with Triton X-114 indicated that suPAR associates with the aqueous phase, indicative of the absence of the glycolipid anchor. There was a polarized release of suPAR from cultured endothelial cells towards the basolateral direction, whereas the membrane-bound receptor was found preferentially on the apical surface. Both, uPAR and suPAR became upregulated 2-4 fold after activation of protein kinase C with phorbol ester, which required de-novo protein biosynthesis. Interleukin-1beta (IL-1beta), basic fibroblast growth factor (bFGF) or vascular endothelial growth factor increased suPAR release from endothelial cells, whereas platelet derived growth factor-BB, bFGF or IL-1beta stimulated suPAR release from vascular smooth muscle cells. Immune electron microscopy indicated that in atherosclerotic vessels (s)uPAR was observed on cell membranes as well as in the extracellular matrix. These findings indicate that (s)uPAR from vascular cells is upregulated by proangiogenic as well as proatherogenic growth factors and cytokines, is preferentially released towards the basolateral side of endothelial cells and accumulates in the vessel wall.


Asunto(s)
Endotelio Vascular/metabolismo , Activadores Plasminogénicos/metabolismo , Receptores de Superficie Celular/metabolismo , Arteriosclerosis/metabolismo , Arteriosclerosis/patología , Técnicas de Cultivo de Célula , Membrana Celular/química , Polaridad Celular , Medios de Cultivo Condicionados/análisis , Endotelio Vascular/citología , Endotelio Vascular/efectos de los fármacos , Sustancias de Crecimiento/farmacología , Humanos , Microscopía Electrónica , Músculo Liso Vascular/citología , Músculo Liso Vascular/efectos de los fármacos , Músculo Liso Vascular/metabolismo , Ésteres del Forbol/farmacología , Activadores Plasminogénicos/biosíntesis , Activadores Plasminogénicos/química , Receptores de Superficie Celular/biosíntesis , Receptores de Superficie Celular/química , Receptores del Activador de Plasminógeno Tipo Uroquinasa , Solubilidad , Regulación hacia Arriba/efectos de los fármacos
15.
Curr Opin Cell Biol ; 12(5): 621-8, 2000 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-10978899

RESUMEN

In a variety of cell types, the glycolipid-anchored urokinase receptor (uPAR) is colocalized pericellularly with components of the plasminogen activation system and endocytosis receptors. uPAR is also coexpressed with caveolin and members of the integrin adhesion receptor superfamily. The formation of functional units with these various proteins allows the uPAR to mediate the focused proteolysis required for cell migration and invasion and to contribute both directly and indirectly to cell adhesive processes in a non-proteolytic fashion. This dual activity, together with the initiation of signal transduction pathways by uPAR, is believed to influence cellular behaviour in angiogenesis, inflammation, wound repair and tumor progression/metastasis and open up the way for uPAR-based therapeutic approaches.


Asunto(s)
Adhesión Celular/fisiología , Receptores de Superficie Celular/fisiología , Animales , Humanos , Integrinas/metabolismo , Unión Proteica , Receptores de Superficie Celular/metabolismo , Receptores del Activador de Plasminógeno Tipo Uroquinasa
16.
Blood ; 96(2): 514-22, 2000 Jul 15.
Artículo en Inglés | MEDLINE | ID: mdl-10887113

RESUMEN

Proteolytic cleavage of single-chain high molecular weight kininogen (HK) by kallikrein releases the short-lived vasodilator bradykinin and leaves behind 2-chain high molecular weight kininogen (HKa) that has been previously reported to exert antiadhesive properties as well as to bind to the urokinase receptor (uPAR) on endothelial cells. In this study we defined the molecular mechanisms for the antiadhesive effects of HKa related to disruption of integrin- and uPAR-mediated cellular interactions. Vitronectin (VN) but not fibrinogen or fibronectin-dependent alphavbeta(3) integrin-mediated adhesion of endothelial cells was blocked by HKa or its isolated domain 5. In a purified system, HKa but not HK competed for the interaction of VN with alphavbeta(3) integrin, because HKa and the isolated domain 5 but not HK bound to both multimeric and native VN in a Zn(2+)-dependent manner. The interaction between HKa or domain 5 with VN was prevented by heparin, plasminogen activator inhibitor-1, and a recombinant glutathione-S-transferase (GST)-fusion peptide GST-VN (1-77) consisting of the amino terminal portion of VN (amino acids 1-77), but not by a cyclic arginyl-glycyl-aspartyl peptide, indicating that HKa interacts with the amino terminal portion of VN ("somatomedin B region"). Furthermore, we have confirmed that HKa but not HK bound to uPAR and to the truncated 2-domain form of uPAR lacking domain 1 in a Zn(2+)-dependent manner. Through these interactions, HKa or its recombinant His-Gly-Lys-rich domain 5 completely inhibited the uPAR-dependent adhesion of myelomonocytic U937 cells and uPAR-transfected BAF-3 cells to VN and thereby promoted cell detachment. By immunogold electron microscopy, both VN and HK/HKa were found to be colocalized in sections from human atherosclerotic coronary artery, indicating that the described interactions are likely to take place in vivo. Taken together, HK and HKa inhibit different VN-responsive adhesion receptor systems and may thereby influence endothelial cell- or leukocyte-related interactions in the vasculature, particularly under inflammatory conditions. (Blood. 2000;96:514-522)


Asunto(s)
Adhesión Celular , Quininógenos/fisiología , Receptores de Superficie Celular/fisiología , Receptores de Vitronectina/fisiología , Arteriosclerosis/metabolismo , Endotelio Vascular/citología , Endotelio Vascular/metabolismo , Fibrinógeno/farmacología , Humanos , Quininógenos/análisis , Quininógenos/metabolismo , Leucocitos/fisiología , Peso Molecular , Monocitos , Osteosarcoma , Inhibidor 1 de Activador Plasminogénico/análisis , Receptores de Superficie Celular/análisis , Receptores del Activador de Plasminógeno Tipo Uroquinasa , Células Tumorales Cultivadas , Células U937 , Vitronectina/análisis , Vitronectina/metabolismo , Vitronectina/farmacología , Zinc/farmacología
17.
J Pathol ; 190(3): 360-72, 2000 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-10685070

RESUMEN

The endothelium plays a crucial dynamic role as a protective interface between blood and the underlying tissues during the haemostatic process, which maintains blood flow in the circulation and prevents life-threatening blood loss. Following vessel wall injury with initial platelet adhesion and aggregation to exposed subendothelial extracellular matrix, the initiation, amplification, and control of haemostasis depend on structurally unrelated membrane-associated receptors for blood coagulation proteases including tissue factor, G-protein-coupled protease-activatable receptors, thrombomodulin, and protein C receptor, respectively. In addition to their regulatory role in haemostasis, the respective (pro-)enzyme ligands such as Factors VIIa and Xa, thrombin or protein C mediate specific signalling pathways in vascular cells related to migration, proliferation or adhesion. The functional importance of these receptors beyond haemostasis has been manifested by various lethal and pathological phenotypes in knock-out mice. These protease receptors thereby provide important molecular links in the vascular system and serve to integrate haemostasis with endothelial cell functions which are relevant for the (patho-)physiological responses to injury or inflammatory challenges.


Asunto(s)
Endopeptidasas/fisiología , Endotelio Vascular/fisiología , Hemostasis/fisiología , Receptores de Superficie Celular/fisiología , Animales , Factores de Coagulación Sanguínea/fisiología , Comunicación Celular/fisiología , División Celular , Movimiento Celular/fisiología , Endotelio Vascular/citología , Humanos , Ratones , Ratones Noqueados , Trombomodulina/fisiología , Tromboplastina/fisiología
18.
Basic Res Cardiol ; 94(5): 315-21, 1999 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-10543306

RESUMEN

Cell-cell and cell-matrix interactions are key events in morphogenetic processes during development and tissue remodelling. In the vascular system, overexpression of adhesion receptors such as integrins, protease (receptors) or dysregulation of adhesive interactions are directly related to the pathophysiology of cardiovascular diseases (atherosclerosis, restenosis, thrombosis) or angiogenesis-driven tumor progression. Protease cascades such as the plasminogen activation system exhibit a dual role in cell invasion by promoting pericellular proteolysis as well as by regulating cell adhesion and migration in a non-proteolytic fashion. In both these mechanisms, the urokinase receptor (uPAR) plays a central role and may become engaged in complexes with beta1-, beta2-, and beta3-integrins. This article will focus on the molecular and functional interactions between the uPAR system and vascular integrins and discuss implications for cardiovascular function.


Asunto(s)
Enfermedades Cardiovasculares/fisiopatología , Fenómenos Fisiológicos Cardiovasculares , Sistema Cardiovascular/fisiopatología , Adhesión Celular/fisiología , Matriz Extracelular/fisiología , Receptores de Superficie Celular/fisiología , Animales , Humanos , Modelos Cardiovasculares , Receptores del Activador de Plasminógeno Tipo Uroquinasa , Activador de Plasminógeno de Tipo Uroquinasa/fisiología
19.
Blood ; 93(9): 2976-83, 1999 May 01.
Artículo en Inglés | MEDLINE | ID: mdl-10216093

RESUMEN

The trace element Zinc (Zn2+) has been implicated as a mediator in host defense, yet the molecular basis for its extracellular functions remains obscure. Here, we demonstrate that Zn2+ can induce the adhesion of myelomonocytic cells to the endothelium, as well as to the provisional matrix proteins vitronectin (VN) and fibrinogen (FBG), which are pivotal steps for the recruitment of leukocytes into inflamed/injured tissue. Physiologic concentrations of Zn2+ increased the urokinase receptor (uPAR)-mediated adhesion of myelomonocytic cells to VN, whereas other divalent cations had smaller effects. Zn2+-induced cell adhesion to VN was abolished by cation chelators such as 1-10-phenanthroline, as well as by plasminogen activator inhibitor-1 (PAI-1) and a monoclonal antibody (MoAb) against uPAR. These characteristics could be recapitulated with a uPAR-transfected cell line emphasizing the specificity of this receptor system for Zn2+-dependent cell adhesion. Like urokinase (uPA), Zn2+ increased the binding of radiolabeled VN to uPAR-expressing cells, as well as the interaction of VN with immobilized uPAR in an isolated system. Moreover, Zn2+ enhanced leukocytic cell adhesion to FBG and endothelial cell monolayers by activating beta2-integrins. Instead of the direct beta2-integrin activation through the divalent cation binding site, Zn2+-induced integrin activation was mediated via uPAR, a crucial regulator of this system. The present study uncovers for the first time Zn2+-mediated cell adhesion mechanisms that may play a crucial role in modulating leukocyte adhesion to vessel wall components.


Asunto(s)
Antígenos CD18/fisiología , Leucocitos/fisiología , Receptores de Superficie Celular/fisiología , Vitronectina/fisiología , Zinc/farmacología , Anticuerpos Monoclonales/farmacología , Cationes Bivalentes/farmacología , Adhesión Celular/efectos de los fármacos , Quelantes/farmacología , Cobalto/farmacología , Cobre/farmacología , Fibrinógeno/fisiología , Humanos , Cinética , Leucocitos/efectos de los fármacos , Manganeso/farmacología , Níquel/farmacología , Fenantrolinas/farmacología , Inhibidor 1 de Activador Plasminogénico/farmacología , Receptores del Activador de Plasminógeno Tipo Uroquinasa , Acetato de Tetradecanoilforbol/farmacología , Células U937 , Activador de Plasminógeno de Tipo Uroquinasa/metabolismo , Activador de Plasminógeno de Tipo Uroquinasa/farmacología
20.
Br J Haematol ; 104(4): 901-8, 1999 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-10192457

RESUMEN

In the present study the ability of plasminogen activator inhibitor type-1 (PAI-1) to interfere with platelet and megakaryoblastic cell adhesion was investigated. Both cell types exhibited integrin-dependent adhesion in a static system, mediated by alphaIIb beta3 on platelets and alpha v-integrins on different megakaryoblastic cell lines, even though they also expressed alphaIIb beta3. In a concentration-dependent manner, active, but not latent or complexed, PAI-1 abrogated cell adhesion onto vitronectin but not onto fibrinogen or other matrix substrata. Urokinase as well as thrombin neutralized the anti-adhesive effect of active PAI-1. The direct binding of vitronectin, but not of other matrix proteins, to integrin alphaIIb beta3 was blocked by active PAI-1 in a purified system. Since activated platelets release active and latent PAI-1 as well as structurally and functionally distinct forms of vitronectin, the described interactions appear to be physiologically significant. Co-distribution of vitronectin and PAI-1 at sites of fibrin polymers within platelet thrombi was demonstrated by transmission electron microscopy, suggesting an extracellular functional relationship of both release products with regard to cell adhesion. Our data emphasize the regulatory role of active PAI-1 in platelet adhesion to provisional matrix proteins as found during wound healing independent of its anti-proteolytic activity. Furthermore, megakaryocyte maturation may depend on the intact vitronectin-integrin adhesion system that is influenced by PAI-1, thereby proposing a regulatory role for the inhibitor in cellular differentiation.


Asunto(s)
Plaquetas/fisiología , Megacariocitos/fisiología , Inhibidor 1 de Activador Plasminogénico/fisiología , Vitronectina/fisiología , Adhesión Celular/fisiología , Fibrina/metabolismo , Fibrinógeno/metabolismo , Humanos , Complejo GPIIb-IIIa de Glicoproteína Plaquetaria/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA